Sei sulla pagina 1di 11

Muley et al

Tropical Journal of Pharmaceutical Research, October 2009; 8 (5): 455-465


© Pharmacotherapy Group,
Faculty of Pharmacy, University of Benin,
Benin City, 300001 Nigeria.
.
All rights reserved

Available online at http://www.tjpr.org


Review Article

Phytochemical Constituents and Pharmacological


Activities of Calendula officinalis Linn (Asteraceae): A
Review

BP Muley*, SS Khadabadi and NB Banarase


Govt. College of Pharmacy, Kathora Naka, Amaravati, Maharashtra State, India- 444 604.

Abstract
Calendula officinalis Linn. (Asteraceae) is used medicinally in Europe, China and India amongst several
places in the world. It is also known as “African marigold” and has been a subject of several chemical
and pharmacological studies. It is used in traditional medicine, especially for wound healing, jaundice,
blood purification, and as an antispasmodic. Chemical studies have underlined the presence of various
classes of compounds, the main being triterpenoids, flavonoids, coumarines, quinones, volatile oil,
carotenoids and amino acids. The extract of this plant as well as pure compounds isolated from it, have
been demonstrated to possess multiple pharmacological activities such as anti-HIV, cytotoxic, anti-
inflammatory, hepatoprotective, spasmolytic and spasmogenic, amongst others. In this review, we have
explored the phytochemistry and pharmacological activities of C. officinalis in order to collate existing
information on this plant as well as highlight its multi-activity properties as a medicinal agent. This is as a
result of the worldwide cultivation of the plant and increasing published reports on it.

Key words: Calendula officinalis, Asteraceae, Phytochemical constituents, Pharmacological activities,


Contraindications.

Received: 11 April 2009 Revised accepted: 10 September 2009

*Corresponding author: Email: muley.b@gmail.com; nbanarase7@gmail.com.

Trop J Pharm Res, October 2009; 8 (5): 455


Muley et al

INTRODUCTION up of orange-yellow tubular florets. The disc


florets are pseudohermaphrodites but the
India is called the botanical garden of the female is sterile. The zygomorphic ray florets
world for its rich natural resources. Over at the edge are female, their stamens are
6,000 plants in India are used in traditional, completely absent, and their inferior ovaries
folklore and herbal medicine[1]. The Indian are much more developed than those of the
system of medicine has identified 1500 tubular florets. The fruit forms only in the
medicinal plants of which 500 are commonly female ray flowers. The heterocarp achenes
used[1]. Calendula officinalis Linn. is used are sickle-shaped, curved and ringed.
medicinally in Europe, China, US and India. It
belongs to the family, Asteraceae, and is Leaf, stem and root
commonly known as Zergul (Hindi), African
marigold, Calendula, Common Marigold, The plant is an annual, seldom biennial. It
Garden Marigold, Marigold, Pot Marigold grows to between 30 and 50 cm high, and
(English), Butterblume (German), Chin Chan has about 20 cm long tap root and numerous
Ts’ao (Chinese), Galbinele (Romanian) and thin, secondary roots. The stem is erect,
Ringblomma (Swedish)[2,3]. angular, downy and branched from the base
up or higher. The alternate leaves are almost
Taxonomic description spatulate at the base, oblong to lanceolate
above and are all tomentosae[5-7].
The plant is classified as shown in Table 1.

Table 1: Taxonomic classification of Calendula


officinalis [4]

Kingdom Plantae
Subkingdom Tracheobionta
Division Magnoliophyta
Class Magnoliopsida
Subclass Asteridae Leaves with stem Flower
Order Asterales Fig 1: The leaf, stem and flower of C. officinalis
Family Asteraceae
Tribe Calenduleae
Genus Calendula
Traditional uses
Species C. officinalis
In Europe, the leaves are considered
Habitat resolvent and diaphoretic while the flowers
are used as a stimulant, antispasmodic and
The plant is native to Central and Southern emmenagogue[2]. In England, the decoction
Europe, Western Asia and the US [5]. of the flowers was used as a posset drink for
the treatment of measles and smallpox, and
Botanical description the fresh juice as a remedy for jaundice,
Flower and fruit costiveness (constipation) and suppression of
menstrual flow [8]. In India, the florets are
On the tip of each stem, there is a 5 to 7 cm used in ointments for treating wounds,
composite flower head, as shown in Fig 1, herpes, ulcers, frostbite, skin damage, scars
consisting of an epicalyx of numerous and blood purification. The leaves, in
narrow-lanceolate sepals, which are densely infusion, are used for treating varicose veins
covered on both sides with glandular hairs. externally [2,8].
The inner section of the flower head is made

Trop J Pharm Res, October 2009; 8 (5): 456


Muley et al

PHYTOCHEMISTRY neohesperidoside, isorhamnetin-3-O-


G
neohesperidoside, isorhamnetin-3-O-2 -
A number of phytochemical studies have rhamnosyl rutinoside, isorhamnetin-3-O-
demonstrated the presence of several rutinoside, quercetin-3-O-glucoside and
classes of chemical compounds, the main quercetin-3-O-rutinoside[18].
ones being terpenoids, flavonoids,
coumarines, quinones, volatile oil, Coumarins
carotenoids and amino acids.
The ethanol extract of the inflorescence of
Terpenoids the C. officinalis reported to contain
coumarins - scopoletin, umbelliferone and
Various terpenoids (Table 2) have been esculetin [29] .
reported from the petroleum ether extract of
C.officinalis flowers. They include sitosterols, Quinones
stigmasterols[9], diesters of diols[10], 3-
monoesters of taraxasterol, ψ-taraxasterol, Quinones reported from C. officinalis were
lupeol[11,12], erythrodiol, brein[13,14], plastoquinone, phylloquinone, α-tocopherol in
ursadiol[15], faradiol-3-O-palmitate, faradiol- the chloroplast, ubiquinone, phylloquinone, α-
3-O-myristate, faradiol-3-O-laurate[16], tocopherol in mitochondria, and
arnidiol-3-O-palmitate, arnidiol-3-O-myristate, phylloquinone in the leaves [30].
arnidiol-3-O-laurate, calenduladiol-3-O-
palmitate, calenduladiol-3-O-myristate[17,18], Volatile oil
oleanolic acid saponins: calenduloside A-
H[19-22], oleanane triterpene glycoside: C. officinalis flowers contain maximum
calendulaglycoside A, calendulaglycoside volatile oil at full flowering stage (0.97 %) and
A6′-O-n-methyl ester, calendulaglycoside A6’- minimum during the preflowering stage (0.13
O-n-butyl ester, calendulaglycoside B, %) [31]. The composition also showed
calendulaglycoside B 6′-O-n-butyl ester, different patterns at different phases of
calendulaglycoside C, calendulaglycoside C vegetative cycles. Various monoterpenes and
6′-O-n-methyl ester, calendulaglycoside C 6′- sesquiterpenes have been reported in the
O-n-butyl ester, calenduloside F6′-O-n-butyl volatile oil : α-thujene, α-pienene, sabinene,
ester, calnduloside G6′-O-n-methyl ester[18], β-pienene, limonene, 1,8-cineol, p-cymene,
glucosides of oleanolic acid (mainly found in trans-β-ocimene, γ-terpenene, δ-3-carene,
roots of grown and senescing plants) I, II, III, nonanal, terpene-4-ol, 3-cylohexene-1-ol, α-
VI, VII [23,24], and glucuronides (mainly phellandrene, α-terpeneol, geraniol,
found in flowers and green parts) F, D, D2, C, carvacrol, bornyl acetate, sabinyl acetate, α-
B and A[25]. One new triterpenic ester of cubebene, α-copaene, α-bourbonene, β-
olanane series has been isolated from cubebene, α-gurjunene, aromadendrene, β-
flowers was cornulacic acid acetate from caryophyllene, α-ylangene, α-humulene, epi-
flowers [26]. bicyclo-sequiphellandrene, germacrene D,
alloaromadendrene, β-saliene, calarene,
Flavonoids muurolene, δ-cadinene, cadina 1,4-diene, α-
cadinene, nerolidol, palustron,
Various flavonoids (Table 3) have been endobourbonene, oplopenone, α-cadinol, T-
isolated from the ethanol extract of the muurolol. The essential oil was found to be
inflorescence of C. officinalis. They include rich in α-cadinene, α-cadinol, t-muurolol,
quercetin, isorhamnetin[27], isoquercetin, limonene, and 1,8-cineol with p-cymene at
isorhamnetin-3-O-β-D-glycoside, narcissin, lower levels at the post-flowering periods
calendoflaside [28], calendoflavoside, [31].
calendoflavobioside, rutin, isoquercitrin,

Trop J Pharm Res, October 2009; 8 (5): 457


Muley et al

Table 2: Structures and activities of some terpenoids

Structure Activity
1 2 3
Calendulaglycoside A: R =Glc, R =Gal, R =H,
4
R =Glc;
1
Calendulaglycoside A6’-O-methyl ester: R =Glc,
2 3 4
R =Gal, R =Me, R =Glc;
1
Calendulaglycoside A6’-O-n-butyl ester: R =Glc,
2 3 4
R = Gal, R =n-Bu, R =Glc;
1 2 3
Calendulaglycoside B: R =Glc, R =Gal, R = H, Responsible for
4
R = H; antitumor, anti-
1
Calendulaglycoside B6’-O-n-butyl ester: R =Glc, inflammatory and
2 3 4
R =Gal, R =n-Bu, R =H; antioedematous
1 2 3
Triterpenoid Calendulaglycoside C: R =H, R =Gal, R =H, activities.
4
R =Glc;
1
Calendulaglycoside C6’-O-methyl ester: R = H,
2 3 4
R =Gal, R =Me, R =Glc;
1
Calendulaglycoside C6’-O-n-butyl ester: R =H,
2 3 4
R =Gal, R =n-Bu, R =Glc;
1
Calendulaglycoside F6’-O-n-butyl ester: R =H,
2 3 4
R =Gal, R =Me, R =H

Responsible for
anti-inflammatory
1 2 3
Faradiol: R =OH, R =R =H and
1 2 3
ψ-Taraxasterol: R =R =R =H antioedematous
activities.

Triterpenoid saponin

Carotenoids violaxanthin, 13Z-violaxanthin, anthera-


xanthin, mutatoxanthin epimer 1,
The methanol extract of leaves, petals and mutatoxanthin epimer 2, lutein, 9/9′ 2-lutein,
pollens of C. officinalis flowers showed a α-cryptoxanthin, β-cryptoxanthin, β-carotene.
number of carotenoids. The carotenoids Total carotenoids (mg/g dry weight) for the
found in the pollens and petals were leaves is 0.85 % and for stems 0.18 %
neoxanthin, 9Z-neoxanthin, violaxanthin, [32,33].
luteoxanthin, auroxanthin, 9Z-violaxanthin,
flavoxanthin, mutatoxanthin, 9Z- Amino acids
anthroxanthin, lutein, 9/9′A-lutein, 13/13′Z-
lutein, α-cryptoxanthin, β-cryptoxanthin, z- The ethanol extract of the flowers of the plant
cryptoxanthin, lycopene, α-carotene, and β- is reported to show the presence of 15 amino
carotene. Total carotenoids (mg/g dry weight) acids in free form: alanine, arginine, aspartic
was 7.71 % for petals and1.61 % for pollens. acid, aspargine, valine, histidine, glutamic
acid, leucine, lysine, proline, serine, tyrosine,
threonine, methionine and phenylalanine.
Reported carotenoid compositions of the Amino acid content of the leaves is about 5
leaves and stems reported were neoxanthin, %, stems 3.5 % and flowers 4.5 % [34].
9Z-neoxanthin, violaxanthin, luteoxanthin, 9Z-

Trop J Pharm Res, October 2009; 8 (5): 458


Muley et al

Table 3: Structures and activities of some flavonoids

Structure Activity
1 2 3
R =Me, R =Rha, R =H;
G
Isorhamnetin3-O-2 -
1
rhamnosylrutinoside: R =Me, Responsible for
2 3
R =Rha, R =Rha; antioxidant and
Isorhamnetin3-O-rutinoside: wound healing
1 2 3
R =Me, R =H, R =Rha; activities
1
Quercetin-3-O-glucoside: R =
2 3
H, R =H, R =H; Quercetin-3-
1 2
O-rutinoside: R =H, R =H,
3
Isorhamnetin3-O-neohesperidoside R =Rha

Responsible for
antioxidant
activities

Quercetin

Responsible for
antioxidant
activities

Isorhamnetin

Carbohydrates monols, sterol esters, 3-monoesters, 3-


monoester diols reported in flowers were
The ethanol extract of the inflorescence of lauric, myristic, palmitic, stearic, oleic, linoleic
plant showed the presence of and linolenic acid. The fatty acids of marigold
polysaccharides, PS-I,-II, and -III having a seeds contain about 59% of an 18:3
(1→3)-β-D-galactam backbone with short conjugated trienic (trans-8,trans-10, cis-12)
side chains at C-6 comprising α-araban- acid and about 5% of 9-hydroxy-18:2 (trans-
(1→3)-araban and alpha-L-rhamnan-(1→3)- 9,cis-11) acid - dimorphecolic acid [37,38]
araban along with monosaccharides [35,36]. one oxygenated fatty acid also reported from
the seed oil of C. officinalis was D-(+)-9-
Lipids hydroxy-10,12-octadecadienoic acid [39].

The lipids in the petroleum ether extract of Other constituents


the seeds, leaves and flowers of C. officinalis
have been analyzed. The amount of neutral Other phtytochemicals include the bitter
lipids in the seeds was 15.7 %, phospholipids constituent, loliolide (calendin) [40], calendulin
0.6 % and glycolipids 0.9 %. Fatty acids of [41] and n-paraffins [42].

Trop J Pharm Res, October 2009; 8 (5): 459


Muley et al

PHARMACOLOGICAL ACTIVITIES activity in in vitro MTT/tetrazolium-based


assay. This activity was attributed to inhibition
Pharmacological studies have confirmed that of HIV1-RT at a concentration of 1,000 µg
C.officinalis exhibit a broad range of /mL as well as suppression of the HIV-
biological effects, some of which are very mediated fusion at 500 µg/mL [47].
interesting for possible future development.
Antibacterial and antifungal activities
Anti-inflammatory and antioedematous
activities The methanol extract and 10 % decoction of
the plant’s flowers were assessed for their
Ethyl acetate soluble fraction of the methanol activity against anaerobic and facultative
extract of C. officinalis flowers exhibited the aerobic periodontal bacteria, namely,
most potent inhibition (84 %) of 12-o- Porphyromonos gingivalis, Prevotella spp.,
tetradecanoyl phorbol-13-acetate (TPA)- Furobacterium nucleatum, Caphocytophaga
induced inflammation (1 µg/ear) in mice with gingivalis, Veilonella parvula, Eikenella
an ID50 value of 0.05 - 0.20 mg/ear compared corrodens, Peptostreptococcus micros and
with indomethacin as reference drug. Actinomyces odontolyticus. The results
Furthermore, activity-guided isolation showed showed marked inhibition against all tested
that its activity was mainly due to oleanane- microorganisms with MIC ≥2048 mg/L [48].
type triterpene glycoside[18]. A dose of 1200 When the essential oil of the flowers was
µg/ear of an aqueous-ethanol extract showed tested (using disc diffusion technique) against
20 % inhibition in croton oil-induced mouse various fungal strains, namely, Candida
oedema. The activity was attributed to the albicans(ATCC64548), Candida dubliniensis
presence of triterpenoids, the three most (ATCC777), Candida parapsilosis
active compounds of which were the esters of (ATCC22019), Candida
faradiol-3-myristic acid, faradiol-3-palmitic glabrata(ATCC90030), Candida krusei
acid and 4-taraxasterol [43,44]. (ATCC6258), and yeast isolated from
humans, viz, Candida albicans, Candida
Dichloromethane extract of the plant’s flower dubliniensis, Candida parapsilosis, Candida
heads inhibited croton oil-induced oedema, glabrata, Candida tropicalis, Candida
and further isolation showed that the esters of guilliermondii, Candida krusei and
faradiol-myristic acid, faradiol-palmitic acid Rhodotorella spp., it showed good potential
and ψ-taraxasterol had antioedematous antifungal activity (at 15 µl/disc) [49].
activity with an oedema inhibition of nearly 50
2 Anticancer and lymphocyte activation
% at a dose of 240 µg/cm . Furthermore,
when the doses of these two faradiol esters dual activities
were doubled, oedema inhibition increased to
65 and 66 %, respectively, without any The ethyal acetate soluble fraction of the
synergism between them [45]. A cream methanol extract of C. officinalis flowers has
containing calendula extract has been shown cytotoxic activity in vitro [18]. Further
reported to be effective in dextran and burn activity-guided isolation of that fraction
oedemas as well as in acute lymphoedema in showed that the active compounds were:
rats. Activity against lymphoedema was calenduloside F6'-O-n-butyl ester, which is
primarily attributed to enhancement of active against leukaemia (MOLT-4 and RPMI
macrophage proteolytic activity [46]. 8226), colon cancer (HCC-2998) and
melanoma (LOXIMVI, SK-MEL-5 and UACC-
Anti-HIV activity 62)] cell lines with GI50 values of 0.77-0.99
µmole, except for leukaemia (CCRF-CEM,
Dichloromethane-methanol (1:1) extract of C. GI50 = 23.1 µmole), renal cancer (AK-1, 17.2
officinalis flowers exhibited potent anti-HIV µmole; UO-31, 12.7 µmole) and breast

Trop J Pharm Res, October 2009; 8 (5): 460


Muley et al

cancer (NCI/ADR-RES, >50 µmole)] cell an inhibitory effect of 25 – 26 % at a dose of


lines; and calenduloside G6'-O-methyl ester, 2000 µg/mL [53].
which is active against all the cancer cell
lines mentioned above with GI50≤ 20 µmole Immunostimulant activity
except for ovarian cancer (IGROVI, GI50 =
20.1 µmole) and renal cancer (VO-31, 33.3 The polysaccharide fraction of C. officinalis
µmole) cell lines[18]. Aqueous laser-activated extract showed immunostimulant activity,
calendula flower extract (LACE) showed based on in vitro granulocyte test.
potent in vitro inhibition of tumour cell Polysaccharide III showed the highest
proliferation when assayed against a wide phagocytosis (54 – 100 %) at a concentration
-5 -6
variety of human and murine tumour cell of 10 - 10 mg/mL, while PS-I and PS-II
lines. The inhibition ranged from 70 – 100 % exhibited 40 – 57 and 20 – 30 %
with an IC50 concentration of 60 µg/mL. The phagocytosis, respectively [35,36].
mechanisms of the inhibition were identified
as cell cycle arrest in G0/G1 phase and Antioxidant activity
caspase-3 induced apoptosis. On the other
hand, when LACE was assayed against A 70 % methanol extract of the plant was
human peripheral blood lymphocyte (PBLs) successively extracted with ether, chloroform,
and human natural killer cell lines (NKL) it ethyl acetate and n-butanol leaving a residual
showed in vitro induction of proliferation and aqueous extract which was assayed for
activation of these cells, mainly B- antioxidant activity by liposomal lipid
+ 2+
lymphocytes, CD4 , T lymphocytes and NKT peroxidation-induced Fe and ascorbic acid.
lymphocyte[50]. The ether, butanol and water extracts,
containing flavonoids, showed antioxidant
Various extracts of the leaf, flower and whole activity [54]. Propylene glycol extracts of the
plant have also been found to be cytotoxic to petals and flower heads, assayed for
MRC5, HeP2, ascetic cells from Ehrlich antioxidant activity by lipid peroxidation,
carcinoma. The saponin rich fraction of these indicate that the extract of the petals was
extracts displayed antitumoural activity in vivo more potent than the flower head extract,
in the Ehrlich mouse carcinoma model [51]. based on analysis of plasma and urine
malondialdehyde (MDA) and urine
Hepatoprotective activity isoprostane inventrations (ipf2α-VI)[55].

The hydroalcohol extract of the flowers, when Wound healing activity


given to CCl4-intoxicated liver in albino male
Wistar rats at a dose of 10 mL/kg, resulted in The ethanol extract of the plant’s flowers was
a reduction of hepatocytolysis by 28.5 % due investigated against experimentally induced
to reduction in glutamo-oxalate-transaminase thermal burns in rats. Among the various
(GOT) and glutamo-pyruvate-transaminase extract doses (20, 100, and 200 mg/kg of
(GPT). However, histoenzymology showed body weight), the 200 mg/kg dose showed
reduction of steatosis of lactate significant improvement in healing of wounds
dehydrogenase (LDH), succinate as indicated by increase in collagen-
dehydrogenase (SDH), cytochromoxidase hydroxyproline and hexosamine contents.
2+
(Cyox) and Mg -dependant adenosine The level of acute phase proteins
triphosphatase (ATPase) [52]. The hot water (heptaglobin, orosomycid and tissue damage
extract of C. officinalis flowers exhibited marker enzymes - alkaline phosphatase),
antihepatoma activity against five human liver alanine and aspartate transaminase
cancer cells - HepG2/C3A, SK-HEP-1, decreased significantly. The decrease in lipid
HA22T/VGH, Hep3B and PLC/PRF/5 - with peroxidation might be due to its antioxidant
property [56].

Trop J Pharm Res, October 2009; 8 (5): 461


Muley et al

AEE extracts showed complete reversal of


The daily application of 2% calendula gel DEN effect at levels of around 50 ng/mL, and
resulted in a greater number of wound between 0.4 and 16 ng/mL, respectively. In
healing due to its antimicrobial and the absence of DEN, these two polar extracts
antioxidant property [57]. induced UDS at concentrations of 25 and 3.7
- 100 µg /mL for AE and AEE, respectively, in
Spasmolytic and spasmogenic dual rat liver cell culture. Thus these polar extracts
activity (AE and AEE) at low concentrations (i.e.,
ng/mL range) showed antigenotoxic effect
The aqueous-ethanol extract of C. officinalis
while at high concentrations (i.e., µg/mL
flowers ,when assayed in rabbit jejunum,
range) they exhibited genotoxic effect [61].
caused a dose-dependant (0.03 - 3.0 mg/mL)
+ The propylene glycol extract of C. officinalis
relaxation of spontaneous and K - induced
also showed antigenotoxic effect based on an
contraction; further fractionation of the extract
with dichloromethane showed inhibition of evaluation in young growing pigs which
spontaneous contractions in a dose range of involved the measurement of the excretion of
0.01 - 0.3 mg/mL. This is ten times more lymphocyte DNA fragmentation and 24 h
potent than the parent crude extract, and urinary 8-hydroxy-2′-deoxyguanosine (8-
spasmolytic activity was found to be due to OHdG) [55].
calcium channel blockade (CCB)[58]. On the
other hand, the aqueous fraction of the Antiviral activity
parent extract exhibited spasmogenic activity
in a dose range of 1 - 10 mg/mL [58]. A tincture of the flowers suppressed the
replication of herpes simplex, influenza A2
Insecticidal activity and influenza APR-8 viruses in vitro [62].

The acetone: methanol (2:1 v/v) extract of the TOXOCOLOGICAL STUDY


flowers showed insecticidal activity when it
was tested on milkweedbug[59]. The hydroalcohol extract of C. officinalis
flowers,, based on assessment in rats and
Inhibition of heart rate mice, did not show acute toxicity following
administration of an oral dose of up to 5.0
The aqueous extract was tested on the heart g/kg. It didn’t show haematological alterations
of male Wistar rats and found to inhibit heart at doses of 0.025, 0.25, 0.5 and 1.0 g/kg.
rate contractility by up to 100 % at a dose of
However, the biochemical parameters, blood
0.3mg/L [60].
urea nitrogen (BUN) and alanine
transaminase (ALT), were elevated due to
Genotoxic and antigenotoxic dual
activities renal and liver overload [63].

The aqueous (AE), aqueous-ethanol (AEE), CONTRAINDICATION


ethanol and chloroform extracts of C.
officinalis flowers were evaluated to The extract was found to cause allergy in 9
determine if they caused induction of patients out of 443 (2.03 %) when assessed
unscheduled DNA synthesis (UDS) in rat liver by patch testing method[64]. Therefore, it is
culture and reversal of diethylnitrosamine advisable that the persons who have an
(DEN)-induced UDS. In the UDS test in liver established allergy to the Asteraceae (daisy)
culture, DEN, at a level of 1.25 µmole, family should use it with caution [65,66] .
3
produced a maximum increase of 40% H-
3
thymidine ( HdTT) incorporation while AE and

Trop J Pharm Res, October 2009; 8 (5): 462


Muley et al

CONCLUSION 8. Khare CP. Encyclopedia of Indian Medicinal Plants.


Germany, Springer-Verlag Publisher, 2004, pp
116-117.
In this review, we have presented information 9. Adler G, Kasprzyk Z. Free sterols, steryl esters,
on the botanical description, traditional uses, glycosides, acelyted glycosides and water-
soluble complexes in Calendula officinalis.
phytochemistry and pharmacology of C.
Phytochemistry, 1975; 14: 627-631.
officinalis Linn. (Asteraceae), a medicinal 10. Wilkomirski B, Kasprzyk Z. Free and ester-bound
plant found in central and southern Europe, triterpene alcohols and sterols in cellular
western Asia and the United States, amongst subfractions of Calendula officinalis.
Phytochemistry, 1979; 18: 253-255.
others. A variety of phytochemicals such as 11. Wilkomirski B. Pentacyclic triterpene triols from
terpenoids, flavonoids, coumarins, quinones, calendula officinalis flowers. Phytochemistry,
volatile oil, carotenoids and others have been 1985; 24(12): 3066-3067.
reported to be present in this plant. It exhibits 12. Zittwel-Eglseer K, Sosa S, Jurenitsch J, Schubert-
Zsilavecz M, Loggia RD, Tubaro A, Bertoldi M,
several pharmacological activities such anti-
Franz C. Antioedematous activities of the main
HIV, anti-cancer (dual activity), anti- triterpenoid esters of marigold (Calendula
inflammatory, hepatoprotective, spasmolytic officinalis L.). J Ethnopharmacol, 1997; 57:
and spasmogenic (dual activity), amongst 139-144.
13. Wojciechowski Z, Bochenska-Hryniewicz M,
others. It is potentially an important medicinal
Kurcharezak B, Kasprzyk Z. Sterol and
plant for mankind. triterpene alcohol esters from calendula
officinalis. Phytochemistry, 1972; 11: 1165-
ACKNOWLEDGEMENT 1168.
14. Kasprzyk Z, Wilkomirski B. Structure of a new
triterpene triol from calendula officinalis
The authors wish to thank Professor UA flowers. Phytochemistry, 1973; 12: 2299-2300.
Deokate for the photographs used in this 15. Sliwowski J, Dziewanowska K, Kasprzyk E.
Ursadiol: a new triterpene diol from calendula
review, BA Baviskar for assistance with
officinalis flowers. Khim Prir Soed, 1973; 12:
software used in drawing the chemical 157-160.
structures, and Professor IA Farooqui for 16. Eitterl-Eglseer K, Reznicek G, Jurenitsch J , Novak
financial assistance. J, Zitterl W, Franz C. Morphogenetic variability
of faradiol monoesters in marigold Calendula
officinalis L. Phytochem Anal, 2001; 12: 199-
REFERENCES 201.
17. Neukiron H, D’Ambrosio M, Dovia J, Guerriero A.
1. Agrawal OP, Raju PS. Global market of herbal Simultaneous Quantitative Determination of
products: Opportunities for Indian Traditional Eight Triterpenoid Monoesters from Flowers of
System of Medicine. New Delhi, India, 10 Varieties of Calendula officinalis L. and
Narcosa Publishing House, 2006, pp 5-10. Characterisation of A New Triterpenoid
2. Lt. Colonel Kirtikar KR, Major Basu BD. Indian Monoester. Phytochem Anal, 2004; 15: 30-35.
Medicinal Plants. Vol II, Deharadun, India, 18. Ukiya M, Akihisa T, Yasukava K, Tokuda H, Suzuki
International Book Distributor, 1993, pp 1413- T, Kimura Y. Antiinflammatory, anti-Tumor-
1414. Promoting and Cytotoxic Activities of
3. The Wealth of India, Raw Materials, A Dictionary of Constituents of Marigold (Calendula officinalis)
Indian Raw Material & Industrial Products. Vol Flowers. J Nat Prod, 2006; 69: 1692-1696.
3, New Delhi, Publications & Information 19. Vecherko LP, Sviridov AF, Zinkevich EP, Kogan
Directorate CSIR, 1992, pp 55-58. LM. Structures of calenduloside g and h from
4. Calendula officinalis. From Wikipedia, the free the roots of calendula officinalis. Khim Prir
encyclopedia. [Cited 2009 January 30} Soed, 1974; 4:532-534.
http://en.wikipedia.org/wiki/Calendula_officinali 20. Vecherko LP, Sviridov AF, Zinkevich EP, Kogan
s. LM. The structure of calenduloside C and D
5. Editorial Boards. PDR for Herbal Medicines. 2nd from the roots of Calendula officinalis. Khim
edn, Montvale, Thomson-Medical Economics, Prir Soed, 1975; 3:366-73.
2003; pp 497-500. 21. Vecherko LP, Kabanov US, Zinkevich EP, Kogan
6. Norman GB, Max W. Herbal Drugs & LM. The structure of calenduloside B from the
Phytopharmaceuticals. 2nd edn, Germany, roots of Calendula officinalis. Khim Prir Soed,
MedPharm GmbH Scientific Publishers, 2001; 1971; 4: 533-533.
pp 118-120. 22. Vecherko LP, Zinkevich EP, Libizov NI, Ban’kooskii
7. Ben-Erik VW, Michael W. Medicinal Plants of the AI. Calenduloside A from Calendula officinalis.
Worlds. Times Edition, 2004, pp 74. Khim Prir Soed, 1969; 5(1): 58-59.

Trop J Pharm Res, October 2009; 8 (5): 463


Muley et al

23. Ruszkowski D, Szakiel A, Janiszowska W. 39. Badami RC, Morris LJ. The oxygenated fatty acid of
Metabolism of [3-3H] oleanolic acid in calendula seeds oil. The Journal of the
Calendula officinalis L. roots. APP, 2003; American Oil Chemist’s Society, 1965; 42:
25(4): 311-317. 1119-1121.
24. Wojciechowski Z, Jelonkiewicz-Konador A, 40. Willuhn G, Westhaus RG. Loliolide (Calendin) from
Tomaszewski M, Jankowski J, Kasprzyk Z. Calendula officinalis. Planta Med, 1987; 53:
The structure of glucosides of oleanolic acid 304-304.
isolated from the roots of calendula officinalis 41. Fleisonner AM. Plant extracts: To accelerate
flowers. Phytochemistry, 1971; 10: 1121-24. healing and reduce inflammation. Cosmet
25. Vidal-Ollivier E, Balansard G. Revised structures of Toilet, 1985; 45: 100-113.
triterpenoid saponins from the flowers of 42. Komoe H, Hayashi N. Paraffins of the petals of
Calendula officinalis. J Nat Prod, 1989; 52(5): Calendula officinalis. Phytochemistry, 1971;
1156-1159. 10: 1944-1944.
26. Naved T, Ansari SH, Mukhtar HM, Ali M. New 43. Della LR. Topical anti-inflammatory activity of
triterpenic esters of oleanene-series from the Calendula officinalis extracts. Planta Med,
flowers of Calendula officinalis Linn. Org Chem 1990; 56: 658-658.
Incl Med Chem, 2005; 44: 1088-1091. 44. Della LR, Della LR, Tubaro A, Sosa S, Becker H,
27. Kurkin VA and Sharova OV. Flavonoids from Saar S, Isaac O. The role of triterpenoids in
calendula officinalis flowers. Khim Prir Soed, topical anti-inflammatory activity of Calendula
2007; 2: 179-180. officinalis flowers. Planta Med, 1994; 60: 516-
28. Vidal-Ollivier E. Flavonol glycosides from Calendula 520.
officinalis flowers. Planta Med, 1989; 55: 73- 45. Zitterl-Eglseer K. Anti-oedematous activities of the
73. main triterpenoidal esters of marigold
29. Kerkach AI, Komissarenko NF and Chernobai VT. (Calendula officinalis L.) J Ethnopharmacol,
Coumarines of the inflorescences of Calendula 1997; 57: 139-144.
officinalis and Helichrysum arenarium. Khim 46. Casley-Smith JR. The effects of ‘Unguentum
Prir Soed, 1986; 6: 777-777. lyphaticum’ on acute experimental
30. Janiszowska W, Michalski W, Kasprzyk Z. lymphedema and other high-protein edema.
Polyprenyl quinones and α-tocopherol in Lymphology, 1983; 16: 150-156.
calendula officinalis. Phytochemistry, 1976; 15: 47. Kalvatene Z, Walder R, Gabzaro D. Anti-HIV activity
125-127. of extracts from Calendula officinalis flowers.
31. Okoh OO, Sadimenko AA, Afolayan AJ. The effects Biomed & Pharmacother, 1997; 51: 176-180.
of age on the yield and composition of the 48. Iauk L, Lo-Bue AM, Milazzo I, Rapisarda A,
essential oils of Calendula officinalis. J Appl Blandino G. Antibacterial Activity of Medicinal
Sci; 2007; 7(23): 3806-3810. Plant Extracts Against Periodontopathic
32. Bako E, Deli J, Toth G. HPLC study on the Bacteria. Phytother Res, 2003; 17: 599-604.
carotenoid composition of Calendula products. 49. Gazim ZC, Rezende CM, Fraga SR, Svidzinski TE,
J Biochem Biophys Methods, 2002; 53: 241- Cortez DG. Antifungal activity of the essential
250. oil from calendula officinalis l. (asteraceae)
33. Goodwin TW. Studies in carotenogenesis: the growing in brazil. Braz. J. Microbiol 2008; 39:
carotenoids of the flower petals of calendula 61-63.
officinalis. Biochem J, 1954; 58: 90-94. 50. Medina EJ, Lora AG, Paco L, Algarra I, Collado A,
34. Abajova RL, Aslanov SM, Mamedova ME. Amino Garrido F. A new extract of the plant
acids of calendula officinalis. Chemistry of Calendula officinalis produces a dual in vitro
Natural Compounds, 1994; 30(15): 641-641. effect: cytotoxic antitumor activity and
35. Varlijen J. Structural analysis of lymphocyte activation. BMC Cancer, 2006; 6:
rhamnoarabinogalactans and 119-132.
arabinogalactans with immunostimulating 51. Boucard-Maitre Y, Boucard-Maitre Y, Algernon O,
activity from Calendula officinalis. Raynaurd, J. Genotoxic and antitumoral
Phytochemistry, 1989; 28: 2379-2383. activity of Calendula officinalis extracts.
36. Wagner H, Proksch A, Riess-Maurer I, Vollmar A, Pharmazie, 1988; 43: 220-221.
S. Odenthal, Stuppner H , Jurcic K, Le Turdu 52. Rasu MA, Tamas M, Puica C, Roman I, Sabadas
M, Fang Jn.. Immunstimulierend wirkende M. The hepatoprotective action of ten herbal
Polysaccharide (Heteroglykane) aus höheren extracts in CCl4 intoxicated liver. Phytother
Pflanzen. Arzneimittel-Forschung, 1985; 7: Res, 2005; 19: 744-749.
1069–1075. 53. Lin LT, Liu LT, Chiang LC, Lin CC. In vitro anti-
37. Vlchenko NT, Glushenkova AI and Mukhamedova hepatoma activity of fifteen natural medicines
KS. Lipids of calendula officinalis. Chemistry of from Canada. Phytother Res, 2002; 16: 440-
Natural Compounds, 1998; 34(3): 272-274. 444.
38. Wilkomirski B, Kasprzyk Z. Free and ester-bound 54. Popovic M, Kaurinovic B, Mimica-Dukic N,
triterpene alcohols and sterols in calendula Vojinovic-Miloradov M, Cupic V.. Combined
flowers. Phytochemistry, 1979; 18: 253-255. effects of plant extracts and xenobiotics on
liposomal lipid peroxidation. Part 1. Marigold

Trop J Pharm Res, October 2009; 8 (5): 464


Muley et al

extract-ciprofloxacin/pyralene. Oxidation 61. Perez-Carreon JI, Cryz-Jimener G, Licea-Vega JA,


Commum, 1999; 22: 487-494. Popoca EA, Fazenda SF, Villa-Trevinos.
55. Frankic T, Salobir K, Salobir J. The comparison of Genotoxic and antigenotoxic properties of
in vivo antigenotoxic antioxidative capacity of Calendula officinalis extracts in rat liver culture
two propylene glycol extracts of Calendula treated with diethylnitrosamine. Toxicology in
officinalis (Marigold) and vitamin E in young vitro, 2002; 16: 253-258.
growing pigs. J Anim Physiol Anim Nutr, 2008; 62. Bogdanova NS, Nikolaeva IS, Shcherbakova LI,
41: 1-7. Tolstova TI, Moskalenko NI, Pershin GN.
56. Chandran PK, Kutton R. Effect of Calendula Study of antiviral properties of Calendula
officinalis flower extract on acute phase officinalis. Farmakol Toksikol (Moscow), 1970,
proteins, antioxidant defense mechanism and 33: 349.
granuloma formation during thermal burns. J 63. Silva ER, Goncalves ES, Aguiar F, Evencio LB,
Clin Biochem Nutr, 2008; 43: 58-64. Lyra MA, Coelno MC, Fraga MA, Wanderloy
57. Leach MJ. Calendula officinalis and wound healing: AG. Toxicological studies on hydroalcoholic
A systematic review. Wounds, 2008; 20(8): 1- extract of Calendula officinalis L. Phytother
7. Res, 2007; 21: 332-336.
58. Bashir S, Janbaz KH, Jabeen Q and Gilani AH. 64. Reider N, Comericki P, Hausen BM, Fritsch P,
Studies on Spasmogenic and Spasmolytic Aberer W. The seamy side of natural
Activities of Calendula officinalis flowers. medicines: Contact sensitization to arnica
Phytother Res, 2006; 20: 906-910. (Arnica montana L.) and marigold (Calendula
59. Alexenizor M, Dorn A. Screening of medicinal and officinalis L.). Contact Dermatitis, 2001; 45:
ornamental plants for insecticidal and growth 269-272.
regulating activity. J Pestic. Sci, 2007; 80: 205- 65. Braun L, Cohen M. Herbs and natural supplements:
215. an evidence based guide. Sydney, Elsevier,
60. Perez-Guitierrez S, Vargas-Solis R, Miguel ZS, 2005, pp 98-100.
Perez-G C, Perez-G RM. Inhibitory effect of 66. Bone K. A clinical guide to blending liquid herbs.
five plant extracts on heart rates of rats. St.Louis Missouri, Churchill Livingstone, 2003,
Phytother Res, 1998; 12: S49-50. pp 120-123.

Trop J Pharm Res, October 2009; 8 (5): 465

Potrebbero piacerti anche